Abstract
Increased exposure to antibiotics during early childhood increases the risk of antibiotic-associated dysbiosis, which is associated with reduced diversity of gut microbial species and abundance of certain taxa, disruption of host immunity, and the emergence of antibiotic-resistant microbes. The disruption of gut microbiota and host immunity in early life is linked to the development of immune-related and metabolic disorders later in life. Antibiotic administration in populations predisposed to gut microbiota dysbiosis, such as newborns, obese children, and children with allergic rhinitis and recurrent infections; changes microbial composition and diversity; exacerbating dysbiosis and resulting in negative health outcomes. Antibiotic-associated diarrhea (AAD), Clostridiodes difficile-associated diarrhea (CDAD), and Helicobacter pylori infection are all short-term consequences of antibiotic treatment that persist from a few weeks to months. Changes in gut microbiota, which persist even 2 years after antibiotic exposure, and the development of obesity, allergies, and asthma are among the long-term consequences. Probiotic bacteria and dietary supplements can potentially prevent or reverse antibiotic-associated gut microbiota dysbiosis. Probiotics have been demonstrated in clinical studies to help prevent AAD and, to a lesser extent, CDAD, as well as to improve H pylori eradication rates. In the Indian setting, probiotics (Saccharomyces boulardii and Bacillus clausii) have been shown to reduce the duration and frequency of acute diarrhea in children. Antibiotics may exaggerate the consequences of gut microbiota dysbiosis in vulnerable populations already affected by the condition. Therefore, prudent use of antibiotics among neonates and young children is critical to prevent the detrimental effects on gut health.
Keywords: antibiotic-associated diarrhea, antibiotic-associated dysbiosis, antibiotic resistance genes, Clostridiodes difficile-associated diarrhea, gut microbiota, gut microbiota dysbiosis, Helicobacter pylori infection, probiotics
Introduction
The human gut microbiota (GM) refers to a colony of bacteria, fungi, viruses, and protozoa that dwell in the gastrointestinal (GI) tract of humans.1,2 The human GM is an intricate ecosystem, wherein the resident microorganisms interact among themselves and with the human host.3 The human GM comprises a massive microbial population that plays a crucial role in numerous aspects of human health and survival such as maintenance of host intestinal homeostasis and energy homeostasis, development and modulation of the immune system, metabolic functions, and protection against pathogen colonization in the gut.3,4
Given the significance of GM in health and disease, this review examines antibiotic-associated dysbiosis and its evidence-based role in the most vulnerable populations, which include neonates and children with obesity, allergic rhinitis, or recurrent infections. It also provides a brief overview of the short- and long-term effects of antibiotic-induced microbiota changes, as well as their economic impact. Furthermore, the management of antibiotic-associated dysbiosis has been discussed with a special focus on the role of probiotics in gut dysbiosis and related conditions.
GM dysbiosis
Disturbances in the symbiotic balance between the host and gut microbiota impair intestinal homeostasis and cause GM dysbiosis.1,2,4,5 In this context, GM dysbiosis is defined as a change in the diversity, composition, and function of the gut microbiota.1,2,4 These alterations in the microbiota can be caused by factors related to the host and/or environment and include unbalanced diet, exposure to pathogens and toxins, long-term use of proton pump inhibitors (PPIs), exposure to antibiotics, excessive alcohol consumption, increased sugar or protein intake, pesticide exposure, poor dental hygiene, and long-term stress.6 The gut microorganisms also communicate with the central nervous system through the brain-gut-microbiota axis. Although the exact mechanisms are not clear, endocrine, neural, and metabolic pathways have been suggested. These interactions are considered important and can impact the intestinal homeostasis.7 Disturbances in GM have been linked to the development of several diseases, including metabolic diseases (obesity, diabetes, and metabolic syndrome), atopic diseases (allergies, allergic rhinitis, and asthma), inflammatory and autoimmune diseases [necrotizing enterocolitis, Crohn’s disease, inflammatory bowel disease (IBD), and irritable bowel syndrome].3,8,9 A meta-analysis revealed disease-associated changes in the human gut microbiome with distinct shift patterns.10 In addition, people with inflammatory and metabolic disorders have been found to have less diversity of bacteria than healthy controls.11
Antibiotic-associated dysbiosis
The most frequently prescribed drugs for infectious diseases are antibiotics.5 While primarily targeting disease-causing bacteria, antibiotics also attack beneficial gut microbes, resulting in altered microbiota composition.12 The GI tract is the organ most sensitive to antibiotics taken orally.6 The intestine has three barriers: a physical barrier made up of intestinal epithelial cells (IEC) connected by interepithelial tight junctions, a secretory barrier composed of mucus and antimicrobial peptides (AMP), and an immunological barrier composed of different immune cell types and biomolecules. In addition to affecting the gut microbiota, antibiotic treatment damages the three intestinal barriers. Antibiotic-induced changes in the composition of the gut microbiota alter the production of mucin, cytokines, and AMP, weakening the IEC barrier.6
Different levels of evidence confirm that the administration of antibiotics results in gut microbiota dysbiosis, which mainly involves a reduction in the diversity of gut microbiota, alterations in the abundance of certain taxa, alterations in gene expression, protein activity, and gut metabolome, compromised colonization resistance to invading harmful bacteria, and the emergence of antibiotic-resistant microbes (Figure 1).1,4,8,12 Furthermore, antibiotic-induced changes in the gut microbiota disrupt host–microbial interactions, resulting in increased infectivity and acute gut infections.6,12
Figure 1.
Gut dysbiosis pathology and vulnerable population.
Antibiotics with a broad spectrum of activity that target several bacteria are widely used. Macrolides, penicillins, and vancomycin are examples of such antibiotics. Notably, not all antibiotics affect the microbiome in the same way. Various antibiotics have different modes of action, thus they may wipe out certain subpopulations of microbiota.4 Different antibiotic classes have distinct effects on the GM depending on the drug’s spectrum, delivery route, dosage, and administration duration, as well as the host’s attributes and condition.6,13,14
Compared with adults, infants are more prone to bacterial infections and sepsis, especially premature infants. Antibiotic exposure during infancy causes dysbiosis, which can have detrimental health outcomes in later life.2,4,15–19 Children with obesity, recurrent infections, and allergic rhinitis require multiple doses of antibiotics to combat infections. Repeated exposure to antibiotics alters microbial composition and diversity, which exacerbates dysbiosis and results in substantial health consequences.2 Populations most vulnerable to antibiotic-associated dysbiosis include neonates (first 1000 days of life), children with obesity, children with allergic rhinitis, and children with recurrent infections.
Evidence-based impact of antibiotic administration on gut microbiota in vulnerable populations
Neonates first 1000 days of life
The GM in infants is acquired during birth and subsequently plays a crucial role in the establishment of gut immunity.20 Neonates, especially underweight and premature infants, are susceptible to bacterial infections, and these infections are treated using antibiotics.4,21 Antibiotics are the most often prescribed drugs in critical care units for neonates.22 Antibiotic exposure in early life has been shown to have negative effects on the microbiota and immune system of infants, making them vulnerable to a variety of disorders, including infections, in later life.4,23 The nature of the effects depends upon the class of antibiotics, dosage, route of administration, duration of treatment, and bioavailability.13,14
The development of the GM is influenced by early antibiotic exposure. Delayed microbiota development after antibiotic usage was most noticeable between the ages of 6 and 12 months in a study involving 2-year-old children.24 Evidence from epidemiological studies indicates that early antibiotic exposure in neonates and infants is associated with changes in gut microbiota composition and metabolic activities.18,25,26 Several studies regarding species diversity revealed a substantial link between antibiotic use and a decrease in gut microbiota diversity in neonates, infants, and children.25,27–31 A systematic review including 48 studies evaluated the effects of antibiotic treatment on the infant microbiome and concluded that exposure to antibiotics was linked to decreased microbial diversity and decreased rates of colonization of beneficial bacteria in the gut.32 Of note, penicillin does not exert the same effect on microbiome composition of children as there were no apparent taxonomic changes in the microbiome of untreated and penicillin-treated infants.25
Antibiotic exposure to premature infants can affect their gut flora.20 According to studies, preterm infants who underwent lengthy antibiotic therapy exhibited reduced gut bacterial diversity and species abundance, as well as an upregulation in the genes for antibiotic resistance.33–35 Prolonged treatment with antibiotics in premature infants can increase the risk of acquiring serious infections, and even death.36
Furthermore, the effects of antibiotic-associated dysbiosis might persist for several years. Several observational studies have demonstrated a link between early or repeated exposure to antibiotics during infancy and a higher risk of obesity,15,19,37–44 allergy, and asthma16,17,45 later in childhood. A recent systematic review and meta-analysis advocated that the risk for developing obesity is 20% higher in children who are exposed to antibiotics.46 Several meta-analyses have found that antibiotic use throughout childhood is linked to an increased risk of childhood obesity in a dose-response manner.47,48 Children with earlier postnatal or recurrent exposures to multiple and broad-spectrum antibiotics during the first years of life had a higher risk of childhood obesity.43,46,49,50
Antibiotic usage during late pregnancy increases the abundance of antibiotic-resistant bacteria with increased expression of antibiotic resistance genes (ARGs) in the gut of pregnant women. During delivery, there is transmission of ARGs from the mother to the newborn.1 Exposure to antibiotics within months of birth results in the growth of antibiotic-resistant microbes and spread of antibiotic resistance in the infant gut microbiome.4 According to a study, the incidence of ARGs in the infant GM rises with age and is much higher in infants born by cesarean section.51 Furthermore, ARGs were found on movable genetic elements in infants even after treatment was stopped.31
Overall, infants with antibiotic-driven dysbiosis have altered GM in terms of microbial diversity and abundance, lower resistance to opportunistic pathogens, and increased antibiotic resistance.4,5,8,44
Children with obesity
Antibiotic-associated gut dysbiosis is common in obese people as seen by a decline in the variety and abundance of their gut microbiome.52,53 Generally, the biodiversity depleted due to antibiotic treatment recovers following treatment cessation with significant changes in species composition.2 However, in obese individuals, the microbiome becomes more adept at obtaining energy from various sources as a result of these compositional changes, which affect metabolism and sensitize the host to obesity.3,52
The interplay between gut microbes and obesity is influenced by several metabolic, genetic, and inflammatory mechanisms. The reduced microbial diversity in obese people results in altered levels of metabolites such as succinate and short-chain fatty acids (SCFAs) released from GM-assisted fermentation of indigestible carbohydrates. These metabolites prevent obesity by decreasing appetite and increasing energy consumption.3,54 A study on 42 school children in Malaysia revealed that the total composition of bacterial species was lower in overweight children than in normal-weight children and that there were significant differences in total SCFAs among the two groups.55 Bifidobacteria population during infancy was lower in children becoming overweight, compared with children remaining at a normal weight. It was accordingly suggested that risk of being overweight may be influenced by differences in gut microbiota.56
Studies on children who were exposed to antibiotics during fetal life revealed that obesity and being overweight were linked to early antibiotic administration and were likely to last into adolescent and school-age years even when antibiotics were no longer provided.57 The findings of a study by Schwartz et al.58 suggested that antibiotic administration significantly impacted weight gain irrespective of the age (early or later) at which they are prescribed. Among the different antibiotics, a positive correlation was shown between broad-spectrum antibiotics, such as macrolides, which are active against both aerobic and anaerobic bacteria, and increased body weight.2,59
Children with allergic rhinitis
Allergic rhinitis (AR) is the most common allergic disease that greatly impacts the health of children.60 There are few studies that specifically address the role of gut microbiota in the onset of AR. A cross-sectional study demonstrated a relationship between dysbiosis of specific subgroups of gut microbiota and IgE-mediated reactions to allergens in children aged 4 to 7 years with AR. This could play a role in vulnerability to allergic rhinitis during early childhood.61 A longitudinal prospective study evaluating the changes in gut microbiota composition from early childhood to school age among children diagnosed with IgE-associated allergic diseases (including AR) revealed that Bifidobacterium was abundant, while Lactobacillus, Enterococcus, and Lachnospira were depleted compared with children without IgE-associated allergic diseases.62
As most people with asthma also have AR, the two conditions are commonly linked. The presence of AR increases the probability of developing asthma.63 A systematic literature review reported that the composition of the human GM during the first few months of life was linked to the later development of allergic disease.14 Antibiotic use during the first few days of life has been demonstrated to increase the risk of AR during later stages of childhood.16 A study reported favorable and dose-dependent relationships between early antibiotic exposure (during the first year of life) and the beginning of asthma, and between lifetime antibiotic use and the emergence of AR.17 The findings of this study suggested that while the gut microbiota may become stable and mature within 1 year of birth, it may still be susceptible to insult as the child grows older. Antibiotic insult(s) to the gut microbiota may add up, meaning that the more an infant gets exposed to antibiotics, the more likely he or she will contract the disease (AR and/or asthma) as a child. This suggests that using antibiotics repeatedly can worsen gut dysbiosis.23,64
A Taiwan-based study involving AR patients with and without asthma reported that antibiotic use before the age of three was not linked to the emergence of asthma. However, a 5-year exposure to antibiotics increased the likelihood of developing asthma in a dose-dependent manner. Among children with AR diagnosed with asthma before the age of 12 years, previous 5-year exposure to penicillin and macrolide (but not cephalosporin), markedly increased the likelihood of asthma. However, this was statistically insignificant when asthma was detected after the age of 12 years. In this study, macrolides and penicillin groups were linked to an increased risk of developing asthma among younger children with AR.65
Children with recurrent infections
Infants have an immature microbiome immune system, which increases their risk of recurrent respiratory tract infections (rRTIs).4 Annually, four to eight episodes of rRTIs may be noted in children.66 These infections are treated effectively with the use of antibiotics.67 Antibiotic exposure to infants causes gut microbiota dysbiosis, which in turn alters the innate and adaptive immune responses to pathogens and induces infections associated with a dysfunctional immune system.2,4,13 Antibiotic use during childhood has been linked to a higher incidence of allergies and various infections.68 The use of antibiotics disrupts the communication between the immune system and the microbiota and consequently increases the risk of recurrent infections and allergies.4 A cohort study that investigated the link between an infant’s exposure to antibiotics in the first 6 months of life and the development of eczema, wheezing, and allergic sensitization at 2 years of age found that direct antibiotic exposure increased the likelihood of frequent and persistent wheezing but not of allergic sensitization or eczema.69
Early in life, gut dysbiosis is defined by delayed colonization by anaerobic microbes and increased colonization by opportunistic pathogenic bacteria, especially enterococci and members of the Enterobacteriaceae family.70 In a brief study on children with rRTIs, Li et al.71 found that while the richness of Faecalibacterium, Eubacterium, and Bifidobacterium was much lower in the rRTIs group, Enterococcus was significantly more abundant in this group. In comparison to children who had less than two RTIs in a year, Bosch et al. found that children who had more RTIs in their first year of life had an abnormal pattern of microbial development beginning in the first month of life. These patients had an early abundance of Moraxella, a protracted reduction of Dolosigranulum and Corynebacterium, and reduced stability of the bacterial population.72 Robinson et al.73 reported that Moraxella catarrhalis, Haemophilus influenza, Streptococcus pneumoniae, and Staphylococcus aureus were the most frequently isolated bacteria in children with wheezing.
Short-term and long-term consequences of antibiotic-induced changes in the microbiota of infants and adults. Antibiotics are known to have a negative impact on the composition and diversity of the gut microbiota in adults and children. These effects can be for a short duration or extended periods.1,4,13
Short-term consequences
The effects of antibiotics on gut microbiota composition and diversity can last from weeks to months.4 Antibiotic use throughout childhood leads to a decrease in the diversity and quantity of some microbial species in the gut, particularly Bifidobacterium, as well as low susceptibility to opportunistic infections and increased antibiotic resistance.5 Korpela et al. analyzed feces samples from infants after a course of antibiotics in a recent longitudinal study. They discovered that after a single course of antibiotics, infant gut microbiomes exhibited varying abundances of bifidobacteria, enterobacteria, and clostridia, which lasted for several months. Bifidobacteria levels varied the most in the infant.26 Short-term effects also include antibiotic-associated diarrhea (AAD), Clostridiodes difficile–associated diarrhea (CDAD), and Helicobacter pylori infections.1
Antibiotic-associated diarrhea
Antibiotic-associated diarrhea is a condition caused due to disturbed gut microbiota and stimulated overgrowth of pathogenic or opportunistic strains following administration of antibiotics.74 These organisms mostly include Klebsiella pneumoniae, Staphylococcus aureus, and C difficile.8 The incidence of AAD among children ranges between 11% (outpatient setting) to 21% (inpatients).75 Among adults, it varies between 5% and 70%.76 The prevalence of AAD among hospitalized patients on antibiotics was 9.6%.77 The underlying mechanism of AAD involves antibiotic exposure-driven eradication of gut microbiota and consequent reduction of antimicrobial peptides that control the numbers of intestinal bacteria. Some antibiotics target the mucus layer and tight junctions in the intestinal epithelium, exposing it to more damage. In addition, functions of the mucosal barrier may be affected by alterations in microbial proteases.78 Antibiotics, such as cephalosporins, aminopenicillins without/with clavulanate, clindamycin, and anti-anaerobic antimicrobial agents, whether used orally or intravenously, pose a high risk of causing AAD.79
C difficile–associated diarrhea
In some cases, AAD can be caused by an overgrowth of C difficile, referred to as CDAD.80 It can present as long-term recurrent infections and fatal pseudomembranous colitis.81 Antibiotic use is the main cause of CDAD, and the antibiotics most frequently linked to CDAD are amoxicillin, fluoroquinolones, ampicillin, clindamycin, and cephalosporin.1 Clindamycin has been demonstrated to have a significant unfavorable influence on intestinal microbiota, resulting in decreased resistance to pathogen colonization and an elevated risk of pseudomembranous colitis due to C difficile multiplication.74
H pylori infection
H pylori is a pathogen that inhabits the gastric mucosa. The gastric mucosa becomes inflamed as a result of H pylori infection.1 This reaction is mild and asymptomatic in the majority of instances; however, in some people, it is the primary cause of peptic ulcers and chronic gastritis.1 H pylori infection alters the gastric microbiota, resulting in the development of gastric dysbiosis. These changes are driven by altered pH-gradient across the mucosal surface, leading to the invasion of the gastric mucosa. Mucosal damage leads to increased adhesion and migration of immune cells, which produce inflammatory mediators that cause gastric damage and stop other microorganisms from surviving.82,83 There is a reduction in bacterial diversity in H pylori-infected subjects, with Proteobacteria, Firmicutes, Bacteroidetes, and Actinobacteria being the most prevalent phyla. However, in healthy individuals, gastric microbiota is more diverse and mainly comprises of phyla Bacteroidetes, Firmicutes, Actinobacteria, and Proteobacteria.84
Potential long-term consequences
Antibiotic-induced effects on microbial composition may often last for as long as 2 years after antibiotic exposure. These modifications in the microbiota may result in the development of long-term antibiotic resistance, as well as physiological changes related to the immune system or metabolic function, which remain for years after antibiotic exposure.4 A study analyzed the impact of 7 days of clindamycin treatment on the microbial composition of feces samples and found substantial alterations even 2 years after exposure. Specifically, Bacteroides diversity was lost, abundance of specific taxa was altered, and ARGs were upregulated. These findings show that after short-term exposure to clindamycin, the effects on the composition of the intestinal microbiota may continue for up to 2 years after antibiotic exposure.85 Another study reported that in comparison to the control group that had not been treated with clindamycin, an increased abundance of resistant Bacteroides strains was reported up to 2 years after clindamycin exposure.86 The long-term upregulation of ARG in the human gut is a concerning side-effect of antibiotic treatment.13 Antibiotic exposure during infancy has been linked to a higher chance of acquiring obesity,15,19,37–44 allergies, asthma,16,17,45 and IBD8 later in life.
Economic impact of antibiotic-associated dysbiosis
Antibiotic-associated dysbiosis increases the risk of antibiotic resistance. Resistant bacterial infections increase mortality and morbidity as well as healthcare cost. Antibiotic resistance, according to the Centers for Disease Control and Prevention (CDC), may add nearly US$1400 to hospital bills for treating patients with bacterial infections in the United States alone. This increased expense could rise to more than US$2 billion per year in the future.87 Several studies have shown that antibiotic-associated dysbiosis can be treated with probiotics. However, neither federal medical aid programs nor insurance companies offer coverage for probiotics.88 Antibiotic-associated diarrhea results in higher morbidity, longer stays in the hospital, and greater use of healthcare resources. The increased duration of intensive care unit (ICU) stays and readmission rates result in an additional lifetime cost of £13,272.53 per AAD patient in the United Kingdom. The use of probiotics as a part of a regular perioperative regimen can result in significant cost savings.89 Due to extended and frequent hospitalizations, requirements of medicines, and laboratory tests, CDAD also raises healthcare costs for patients. Healthcare expenses associated with primary CDAD and recurrent CDAD were US$24,205 and US$10,580, respectively, in 2014 in the United States.90 In a prospective study of children aged between 6 and 12 years with acute diarrhea, the direct cost of treatment was INR 779 and INR 944, while the indirect cost was INR 937 and INR 1409 in Group 1 (which received oral rehydration therapy + zinc + Bacillus Clausii as probiotic) and Group 2 (which received oral rehydration therapy + zinc), respectively. The findings indicated that addition of Bacillus Clausii as probiotic helped reduce the duration and frequency of diarrhea, as well as the length of hospital stay, hence lowering the treatment and indirect costs.91
Management of antibiotic-associated dysbiosis
Clinicians should be aware of the potential interaction between gut dysbiosis and metabolic, gastrointestinal, and immunological disorders among children. This can help in ensuring an appropriate treatment approach and also initiating adequate preventive measures. Digestive problems are the main indications of dysbiosis. People with dysbiosis may frequently feel gassy or bloated.92 Dysbiosis is suspected based on clinical symptoms, medical history, physical examination, diagnostic testing (organic acid tests, comprehensive digestive stool analysis, hydrogen breath test, and biopsy), and culture-independent approaches, including next-generation sequencing.93–95 Prebiotics and probiotics may be utilized to treat antibiotic-associated dysbiosis to reduce symptoms and reestablish microbial balance in light of the proven effects of antibiotics on the microbiota.6
Diet supplementation/prebiotics
Diet can quickly and significantly alter the composition of the GM, which will have an impact on the intestinal barrier.96 The GM structure can be modified by a combination of different herbs and vegetables, such as the traditional Chinese herbal medication Shen Ling Bai Zhu San, while receiving antibiotic therapy. It may be beneficial in reducing the presence of harmful bacteria (e.g. Sutterella spp.) while increasing the number of beneficial bacteria (e.g. Bacteroides spp.).97 It was demonstrated in a mouse model that prebiotic fructo-oligosaccharides might undo alterations induced by cefixime in the GM, such as a rise in the number of Akkermansia spp.98 Prebiotics, such as galacto- and short-chain fructo-oligosaccharides, are nondigestible dietary items that stimulate the growth and activity of bacteria, such as Lactobacillus and Bifidobacterium, and restore healthy microbiota following antibiotic exposure.4,99 A 6-g prebiotic supplement containing an inulin-type fructan taken daily prevented antibiotic-induced alterations in the composition of GM by increasing the abundance of Bifidobacterium.100
In a single-center, double-blind, placebo-controlled trial involving overweight/obese children between the ages of 7 and 12, prebiotic oligofructose-enriched inulin (OI) was found to selectively modify the intestinal microbiota and significantly reduce body fat in overweight/obese children.101 Similarly, in a randomized, double-blind, placebo-controlled trial, prebiotic OI supplementation in 7- to 12-year-old obese or overweight children improved appetite control.102 Furthermore, when prebiotics were given to infants in formula-based milk together with antibiotics, the amount of bifidobacteria in their feces was higher than when the children had only received antibiotics.103
Probiotic supplementation
Probiotics are dietary supplements with live microorganisms that have health benefits.4 The microorganisms that are commonly used as probiotics include Lactobacillus species, Bifidobacterium species, Saccharomyces species, and Bacillus species104–107 (Table 1). The probiotic bacteria can modulate intestinal microbiota and can help regulate immune response disorder by stimulating immune cells. The antimicrobial effects of Lactobacilli and Bifidobacteria are attributed to their influence on both local and systemic immunity. Furthermore, they compete with pathogens for nutrients and hence prevent their proliferation. They can also stimulate the release of antimicrobial substances (such as mucin) and prevents adherence of the pathogens to the epithelial barrier.108
Table 1.
Commonly used species as probiotics and their key features.
Probiotic species | Key features |
---|---|
Lactobacillus spp.104 | • A group of lactic acid-producing, nonspore-forming
anaerobes • Considered as ‘friendly’ bacteria taken to recolonize parts of the body to deliver nutritional benefits • Members include Lactobacillus acidophilus, Lactobacillus rhamnosus, Lactobacillus casei, and many more |
Bifidobacterium spp.104 | • A group of lactic acid– and acetic acid–producing
nonspore-forming anaerobes • Used as probiotics in combination with other species • Members include Bifidobacterium brevis, Bifidobacterium infantis, and Bifidobacterium bifidum amongst others |
Saccharomyces spp.104 | • A nonpathogenic yeast strain that has been used to treat
and prevent diarrhea caused by a variety of
sources • Includes Saccharomyces cerevisiae often known as Saccharomyces Boulardii |
Bacillus spp.105–107 | • A group of spore-forming bacteria • Used as probiotics for many decades • Members include Bacillus clausii, Bacillus subtilis, and Bacillus coagulans • Spore-forming bacteria have an advantage over nonspore-forming bacteria such as Lactobacillus spp: ○ B clausii spores are resistant to heat and high temperatures so they can be stored at room temperature with no alterations in their viability. ○ They are also resistant to acidic pH (pH < 7.0) in the stomach and intestine, allowing them to survive passage to the intestine. Of note, B clausii (O/C, SIN, N/R, T) was found alive in the feces for up to 12 days after a single-dose administration of B clausii spores ○ B clausii strains O/C, N/R, SIN, T have been shown to be resistant to at least 14 antibiotics |
Probiotics are shown to alleviate gut dysbiosis and antibiotic-associated side-effects such as diarrhea.109,110 While most studies confirm that probiotics can be effective against gut dysbiosis, more data are needed to ascertain which probiotics are ideal for a specific group of patients, particularly given the wide variations in gut microbiota composition across individuals. Even within a single species, the health advantages of one probiotic strain may not necessarily apply to another.111 Therefore, it is essential to devise new, individualized probiotic supplementation strategies.6 Few next-generation technologies are being evaluated for developing microbiota-related personalized dietary recommendations and the outcomes are promising.112
Role of probiotics
Gut dysbiosis
Probiotics have been shown to confer protection against gut dysbiosis caused by antibiotic treatment. B clausii is the most popular probiotic species that can modify gut microflora and is effective against antibiotic-associated dysbiosis.113 Prolonged supplementation with Lactobacillus rhamnosus GG was reported to alter the intestinal microbiota of children, resulting in a rise in Ruminococcus, Lactococcus, and Prevotella and a decline in Escherichia.114 According to studies, only a few of the species belonging to the genera Bifidobacterium and Lactobacillus are beneficial in terms of their effects on body weight and metabolism.115 A meta-analysis revealed that the use of probiotics appears to be a practical method to reduce the prevalence of RTIs in children.116 According to a randomized trial, younger children who experience recurring respiratory infections over the winter would benefit the most from the use of Lactobacillus GG (LGG).117
Antibiotic-associated diarrhea
There is mounting evidence that probiotic administration can aid in the treatment of AAD as well as the prevention or reduction of the risk of AAD. A multicenter, randomized, open-label, clinical trial that investigated the ability of B clausii to prevent AAD in Filipino infants and children who were given B clausii while receiving antibiotic therapy found that the B clausii-treated group had a decreased incidence of AAD.118 In a longitudinal, multicenter, observational study conducted in Lebanon, an extra course of probiotics to prevent any AAD was given to 118 (48%) of the 246 recruited pediatric participants who were assigned an antibiotic treatment regimen. B clausii was the most commonly recommended probiotic (81.4%; n = 96). Among high-risk patients, the prevalence of diarrhea was twice in the group without probiotics compared with the probiotic group (50% versus 21.9%; versus 50.0% p = 0.182).119 A meta-analysis assessing the effectiveness of probiotics for the prevention of pediatric AAD was undertaken from 1985 to 2013. Overall, probiotics lowered the prevalence of AAD (RR = 0.42, 95% CI: 0.33–0.53) in children (pooled from 22 trials).120 Another meta-analysis of 63 studies showed a statistically significant correlation between probiotic administration and a decline in AAD (p < .001).109 Several studies have shown that the consumption of probiotic combinations orally, including L rhamnosus, Lactobacillus casei, Lactobacillus acidophilus, and Bifidobacterium, lowers the risk of AAD. This effect may be caused by controlling the GM, improving the function of the gut barrier, and modifying immune responses.121–124
Individual or combined strains of Lactobacillus, Bacillus, Lactococcus, Saccharomyces, Bifidobacterium, Leuconostoc cremoris, or Streptococcus spp. have been shown to protect against AAD.125 Clostridium butyricum, Bacillus spp., Lactobacilli spp., Bifidobacterium spp., Lactococcus spp., Saccharomyces spp., L cremoris, or Streptococcus spp., alone or in combination, were evaluated in the Cochrane comprehensive review that included 33 studies involving 6352 subjects. It was concluded that AAD is reduced by probiotics [number needed to treat to benefit (NNTB) 9; 95% CI, 7–13], with large dosages (five billion CFU per day) being preferred.126 According to meta-analyses, certain probiotic strains may help prevent AAD. Therapy with probiotics containing L rhamnosus GG127 or Saccharomyces boulardii128 significantly decreased the risk of AAD in children and adults receiving antibiotics compared with placebo or no therapy. Furthermore, according to the Asian experts’ guidelines on the use of probiotics and B clausii in the management of diarrhea in infants and children, B clausii may be taken into consideration to prevent AAD.129
C difficile–associated diarrhea
Probiotic administration has also been shown to help in the treatment of CDAD as well as the prevention or reduction of the risk of CDAD. A meta-analysis of 20 trials involving 3818 adult or pediatric patients showed that probiotics reduced the incidence of CDAD by 66%.110 A systematic review and meta-analysis of 31 randomized, controlled trials suggested that probiotics can be used to prevent CDAD.130 Furthermore, Saccharomyces boulardii administration decreased the likelihood of CDAD in children (RR, 0.25; 95% CI, 0.08–0.73).131 A meta-analysis of clinical studies assessing probiotics for the prevention of pediatric Clostridiodes difficile infections (CDI) was undertaken from 1985 to 2013. In conclusion, probiotics dramatically decreased CDI in children (pooled from 22 trials).120 In addition, according to the Asian experts’ guidelines on the use of probiotics and B clausii in the management of diarrhea in infants and children, B clausii may be taken into consideration to prevent CDAD.129
H pylori infection
Probiotic strains can potentially increase H pylori eradication rate by 5–10% and are useful in the reduction of related side-effects.132 In a randomized, double-blind, placebo-controlled trial, treatment with B clausii was observed to lessen the frequency of the most prevalent adverse effects (epigastric pain, diarrhea, and nausea) associated with anti-H pylori antibiotic treatment.113 In comparison to the placebo, B clausii capsules reduced the frequency and duration of diarrhea in patients who had H pylori elimination therapy in a randomized, double-blind, placebo-controlled trial. In week 1, the B clausii group had 29% diarrhea and the placebo group had 48% (p < 0.05). In week 2, as compared with placebo, the incidence of diarrhea was still lower with B clausii (20% versus 7.7%) (p < 0.05). From week 1 to week 2, the frequency of days with diarrhea decreased significantly in both groups (p < 0.0001).133 A placebo-controlled, double-blind study found Lactobacillus johnsonii La1 and S boulardii to be effective at reducing H pylori burden.134 Supplementation with L gasseri, L casei DN-114001, L acidophilus, and Bifidobacterium infantis 2036 was linked to significantly greater eradication rates when compared with controls in a meta-analysis of studies in which patients received antibiotic therapy for the eradication of H pylori.135
In addition, the Asian experts’ guidelines on the use of probiotics and B clausii in the management of diarrhea in children suggested that B clausii may be used in conjunction with H pylori abolition therapy.129 According to recent World Gastroenterology Organization (WGO) global guidelines related to H pylori infection, probiotic supplementation may be used to reduce antibiotic-related adverse events.136
Indian perspective on probiotics
It is reported that antibiotics are frequently prescribed to children in India. A study conducted in Bhopal revealed that antimicrobial agents were given to 64% of children under the age of five who were diagnosed with acute gastroenteritis in outpatient facilities.137 Another study conducted in a tertiary referral hospital in Eastern India revealed that 535 antimicrobial drugs were given to 80% of children. In the majority of children (85.6%), antimicrobial therapy was empirical and dependent on clinical judgment.138 Children frequently treated with antibiotics are more vulnerable to gastrointestinal problems, and thus have a special need for probiotic supplementation to maintain a healthy gut flora and a strong immune system. Acute diarrhea is a leading cause of infant and child mortality.139 The studies summarized in Table 2 highlight the use of probiotics in the Indian setting to treat diarrhea in children.
Table 2.
Studies regarding the use of probiotics in the Indian setting to treat diarrhea in children.
First author/ year | Study type | Study population | Findings |
---|---|---|---|
Sudha et al.139 | Double-blind, randomized, placebo-controlled, multicenter study |
N = 119 Children aged 6 months to 5 years with acute diarrhea |
Compared with placebo, B clausii UBBC-07 significantly reduced diarrhea duration (75.66 ± 13.23 versus 81.6 ± 15.43 hours; p < 0.05) and frequency (3.46 ± 0.66 versus 4.57 ± 1.59; p < 0.01), suggesting that the strain is effective in the treatment of pediatric acute diarrhea |
Bhat et al.140 | Open-label randomized controlled trial |
N = 120 Children aged 6 months to 5 years with acute diarrhea |
Duration of diarrhea and hospital stay were reduced in children with acute diarrhea who received Saccharomyces boulardii along with ORT + zinc compared with children who received B clausii + ORT + zinc and ORT + zinc alone |
Vidjeadevan et al.141 | Three-armed randomized controlled trial |
N = 105 Children aged 6 months to 3 years with acute diarrhea |
Adding probiotics such as Saccharomyces boulardii and Bacillus clausii to ORT and zinc helped to shorten the duration of diarrhea |
Das et al.142 | Double-blind randomized controlled trial |
N = 60 Children aged 3 months to 5 years with acute rotavirus diarrhea |
Saccharomyces boulardii was shown to be safe and efficacious in children with acute rotavirus diarrhea by reducing the duration of diarrhea as well as duration of hospitalization |
Lahiri et al.91 | Prospective study |
N = 131 Children aged 6 months to 12 years with acute diarrhea |
Bacillus clausii used as a probiotic along with ORT and zinc reduced the duration (22.64 versus 47.05 hours; p < 0.01), frequency (p < 0.01), and hospital stay (2.78 versus 4.30 days) of diarrhea |
Lahiri et al.143 | Prospective study |
N = 160 Children aged 6 months to 6 years with acute diarrhea |
Bacillus clausii was found to be safe and efficacious with a significant reduction in the duration (22.26 hours versus 34.16 hours; p < 0.05) and frequency of acute diarrhea in children who received probiotic (B clausii) along with ORS + zinc compared with children who received ORS + zinc alone |
ORS, oral rehydration solution; ORT, oral rehydration therapy.
Since the past decade, probiotics have received a lot of attention in India. The introduction of probiotic supplements has coincided with a surge in public awareness primarily in urban areas.144 According to a survey, rising health concerns, increasing stress, changing food habits, and an increase in diabetes, cardiovascular diseases, and lifestyle-related diseases are boosting probiotic demand in India.145 People with special requirements, such as pediatric patients, make up a large portion of India’s probiotic market, and big pharmaceutical companies have been developing probiotic-based nutritional supplements for them.146 The content and analytical validity of probiotics sold commercially in India were rigorously reviewed in a study by Kesavelu et al. There were notable differences in the quantity and quality of probiotics in terms of viable cell count and genus and species content among brand formulations and results of next-generation sequencing.147
Conclusion
Antibiotics can alter the composition and diversity of the gut microbiota in neonates, children, and adults, and may result in antibiotic-associated dysbiosis and intestinal barrier disruption. The disruption of gut microbiota and host immunity in early life, during which the microbiota and immune system are still developing, is linked to the development of immune-mediated and metabolic diseases later in life. Neonates, obese children, and children with allergic rhinitis and recurrent infections comprise the vulnerable populations for antibiotic-associated dysbiosis. The short-term effects of antibiotic use, which last from a few weeks to months, include AAD, CDAD, and H pylori infection. The long-term consequences include changes in gut microbiota, which remain even 2 years after antibiotic exposure, and the development of obesity, allergies, asthma, and IBD. Probiotic bacteria and dietary supplements are highly promising in preventing or restoring the equilibrium disturbed by antibiotics in the gut microbiota. Clinical trials have shown that probiotics can help prevent AAD and, to some extent, CDAD. Probiotics have also been shown to improve H pylori eradication rates. Prescribing antibiotics in vulnerable populations who already present with gut dysbiosis may further exaggerate the effects of dysbiosis. Therefore, it is important to consider the deleterious effects of antibiotics on gut health before prescribing antibiotics to neonates and young children. Prudent use of antimicrobials by following antimicrobial stewardship approaches is the need of the hour and can help prevent gut microbiota dysbiosis to a great extent.
Acknowledgments
The authors acknowledge the BioQuest Solutions Pvt. Ltd., Bangalore, for providing medical writing and editorial support in the preparation of this manuscript paid for by Sanofi India.
Footnotes
ORCID iD: Dhanasekhar Kesavelu
https://orcid.org/0000-0003-4051-3102
Contributor Information
Dhanasekhar Kesavelu, Apollo Children’s Hospital, Chennai 600006, Tamil Nadu, India.
Pramod Jog, Dr. D.Y. Patil Medical College, Hospital & Research Centre, Pune, India.
Declarations
Ethics approval and consent to participate: Not applicable. This article is based on previously conducted studies and does not contain any new data collected from human participants or animals.
Consent for publication: Not applicable.
Author contributions: Dhanasekhar Kesavelu: Conceptualization; Data curation; Formal analysis; Methodology; Resources; Supervision; Validation; Visualization; Writing – review & editing.
Pramod Jog: Conceptualization; Data curation; Formal analysis; Methodology; Resources; Supervision; Validation; Visualization; Writing – review & editing.
Funding: The authors disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This initiative was supported by Sanofi India. Medical writing and open-access related charges were paid for by Sanofi India. The authors received no honoraria from Sanofi directly or indirectly related to the development of this publication.
The authors declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.
Availability of data and material: Data sharing is not applicable to this article as no data sets were generated or analyzed during the current study.
Disclosures: All authors had full access to the articles reviewed in this manuscript, have read and reviewed the final draft of this manuscript, and take complete responsibility for the integrity and accuracy of this manuscript. The content published herein solely represents the views and opinions of the authors. The details published herein are intended for informational, educational, academic, and/or research purposes and are not intended to substitute for professional medical advice, diagnosis or treatment.
References
- 1. Ramirez J, Guarner F, Bustos Fernandez L, et al. Antibiotics as major disruptors of gut microbiota. Front Cell Infect Microbiol 2020; 10: 572912. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Vangay P, Ward T, Gerber JS, et al. Antibiotics, pediatric dysbiosis, and disease. Cell Host Microbe 2015; 17: 553–564. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Chen Y, Zhou J, Wang L. Role and mechanism of gut microbiota in human disease. Front Cell Infect Microbiol 2021; 11: 625913. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Neuman H, Forsythe P, Uzan A, et al. Antibiotics in early life: dysbiosis and the damage done. FEMS Microbiol Rev 2018; 42: 489–499. [DOI] [PubMed] [Google Scholar]
- 5. Parkin K, Christophersen CT, Verhasselt V, et al. Risk factors for gut dysbiosis in early life. Microorganisms 2021; 9: 2066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Duan H, Yu L, Tian F, et al. Antibiotic-induced gut dysbiosis and barrier disruption and the potential protective strategies. Crit Rev Food Sci Nutr 2022; 62: 1427–1452. [DOI] [PubMed] [Google Scholar]
- 7. Karakan T, Ozkul C, Küpeli Akkol E, et al. Gut-brain-microbiota axis: antibiotics and functional gastrointestinal disorders. Nutrients 2021; 13: 389. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Francino MP. Antibiotics and the human gut microbiome: dysbioses and accumulation of resistances. Front Microbiol 2015; 6: 1543. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Biedermann L, Rogler G. The intestinal microbiota: its role in health and disease. Eur J Pediatr 2015; 174: 151–167. [DOI] [PubMed] [Google Scholar]
- 10. Duvallet C, Gibbons SM, Gurry T, et al. Meta-analysis of gut microbiome studies identifies disease-specific and shared responses. Nat Commun 2017; 8: 1784. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Valdes AM. Role of the gut microbiota in nutrition and health. BMJ 2018; 361: j2179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Strati F, Pujolassos M, Burrello C, et al. Antibiotic-associated dysbiosis affects the ability of the gut microbiota to control intestinal inflammation upon fecal microbiota transplantation in experimental colitis models. Microbiome 2021; 9: 39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Jernberg C, Lofmark S, Edlund C, et al. Long-term impacts of antibiotic exposure on the human intestinal microbiota. Microbiology 2010; 156: 3216–3223. [DOI] [PubMed] [Google Scholar]
- 14. Zimmermann P, Curtis N. The effect of antibiotics on the composition of the intestinal microbiota: a systematic review. J Infect 2019; 79: 471–489. [DOI] [PubMed] [Google Scholar]
- 15. Ajslev TA, Andersen CS, Gamborg M, et al. Childhood overweight after establishment of the gut microbiota: the role of delivery mode, pre-pregnancy weight and early administration of antibiotics. Int J Obes 2011; 35: 522–529. [DOI] [PubMed] [Google Scholar]
- 16. Alm B, Goksor E, Pettersson R, et al. Antibiotics in the first week of life is a risk factor for allergic rhinitis at school age. Pediatr Allergy Immunol 2014; 25: 468–472. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Ni J, Friedman H, Boyd BC, et al. Early antibiotic exposure and development of asthma and allergic rhinitis in childhood. BMC Pediatr 2019; 19: 225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Tanaka S, Kobayashi T, Songjinda P, et al. Influence of antibiotic exposure in the early postnatal period on the development of intestinal microbiota. FEMS Immunol Med Microbiol 2009; 56: 80–87. [DOI] [PubMed] [Google Scholar]
- 19. Trasande L, Blustein J, Liu M, et al. Infant antibiotic exposures and early-life body mass. Int J Obes 2013; 37: 16–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Patangia DV, Anthony Ryan C, Dempsey E, et al. Impact of antibiotics on the human microbiome and consequences for host health. Microbiologyopen 2022; 11: e1260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Shane AL, Sanchez PJ, Stoll BJ. Neonatal sepsis. Lancet 2017; 390: 1770–1780. [DOI] [PubMed] [Google Scholar]
- 22. Clark RH, Bloom BT, Spitzer AR, et al. Reported medication use in the neonatal intensive care unit: data from a large national data set. Pediatrics 2006; 117: 1979–1987. [DOI] [PubMed] [Google Scholar]
- 23. Cox LM, Yamanishi S, Sohn J, et al. Altering the intestinal microbiota during a critical developmental window has lasting metabolic consequences. Cell 2014; 158: 705–721. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Bokulich NA, Chung J, Battaglia T, et al. Antibiotics, birth mode, and diet shape microbiome maturation during early life. Sci Transl Med 2016; 8: 343ra82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Korpela K, Salonen A, Virta LJ, et al. Intestinal microbiome is related to lifetime antibiotic use in Finnish pre-school children. Nat Commun 2016; a7: 10410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Korpela K, Salonen A, Saxen H, et al. Antibiotics in early life associate with specific gut microbiota signatures in a prospective longitudinal infant cohort. Pediatr Res 2020; 88: 438–443. [DOI] [PubMed] [Google Scholar]
- 27. Doan T, Arzika AM, Ray KJ, et al. Gut microbial diversity in antibiotic-naive children after systemic antibiotic exposure: a randomized controlled trial. Clin Infect Dis 2017; 64: 1147–1153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Oldenburg CE, Sie A, Coulibaly B, et al. Indirect effect of azithromycin use on the intestinal microbiome diversity of untreated children: a randomized trial. Open Forum Infect Dis 2019; 6: ofz061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Parker EPK, Praharaj I, John J, et al. Changes in the intestinal microbiota following the administration of azithromycin in a randomised placebo-controlled trial among infants in South India. Sci Rep 2017; 7: 9168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Wei S, Mortensen MS, Stokholm J, et al. Short- and long-term impacts of azithromycin treatment on the gut microbiota in children: a double-blind, randomized, placebo-controlled trial. EBioMedicine 2018; 38: 265–272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Yassour M, Vatanen T, Siljander H, et al. Natural history of the infant gut microbiome and impact of antibiotic treatment on bacterial strain diversity and stability. Sci Transl Med 2016; 8: 343ra81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Fjalstad JW, Esaiassen E, Juvet LK, et al. Antibiotic therapy in neonates and impact on gut microbiota and antibiotic resistance development: a systematic review. J Antimicrob Chemother 2018; 73: 569–580. [DOI] [PubMed] [Google Scholar]
- 33. Gasparrini AJ, Wang B, Sun X, et al. Persistent metagenomic signatures of early-life hospitalization and antibiotic treatment in the infant gut microbiota and resistome. Nat Microbiol 2019; 4: 2285–2297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Gibson MK, Wang B, Ahmadi S, et al. Developmental dynamics of the preterm infant gut microbiota and antibiotic resistome. Nat Microbiol 2016; 1: 16024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Greenwood C, Morrow AL, Lagomarcino AJ, et al. Early empiric antibiotic use in preterm infants is associated with lower bacterial diversity and higher relative abundance of enterobacter. J Pediatr 2014; 165: 23–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Esaiassen E, Fjalstad JW, Juvet LK, et al. Antibiotic exposure in neonates and early adverse outcomes: a systematic review and meta-analysis. J Antimicrob Chemother 2017; 72: 1858–1870. [DOI] [PubMed] [Google Scholar]
- 37. Bailey LC, Forrest CB, Zhang P, et al. Association of antibiotics in infancy with early childhood obesity. JAMA Pediatr 2014; 168: 1063–1069. [DOI] [PubMed] [Google Scholar]
- 38. Chelimo C, Camargo CA, Morton SMB, et al. Association of repeated antibiotic exposure up to age 4 years with body mass at age 4.5 years. JAMA Netw Open 2020; 3: e1917577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Leong KSW, McLay J, Derraik JGB, et al. Associations of prenatal and childhood antibiotic exposure with obesity at age 4 years. JAMA Netw Open 2020; 3: e1919681. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Metsala J, Lundqvist A, Virta LJ, et al. Prenatal and post-natal exposure to antibiotics and risk of asthma in childhood. Clin Exp Allergy 2015; 45: 137–145. [DOI] [PubMed] [Google Scholar]
- 41. Scott FI, Horton DB, Mamtani R, et al. Administration of antibiotics to children before age 2 years increases risk for childhood obesity. Gastroenterology 2016; 151: 120–129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Tian J, Liu H, Guo H, et al. Application of antibiotics before 3 years of age increases the risk of childhood overweight and obesity. Exp Ther Med 2021; 21: 56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Baron R, Taye M, Besseling-van der Vaart I, et al. The relationship of prenatal and infant antibiotic exposure with childhood overweight and obesity: a systematic review. J Dev Orig Health Dis 2020; 11: 335–349. [DOI] [PubMed] [Google Scholar]
- 44. Turta O, Rautava S. Antibiotics, obesity and the link to microbes: what are we doing to our children? BMC Med 2016; 14: 57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Yamamoto-Hanada K, Yang L, Narita M, et al. Influence of antibiotic use in early childhood on asthma and allergic diseases at age 5. Ann Allergy Asthma Immunol 2017; 119: 54–58. [DOI] [PubMed] [Google Scholar]
- 46. Duong QA, Pittet LF, Curtis N, et al. Antibiotic exposure and adverse long-term health outcomes in children: a systematic review and meta-analysis. J Infect 2022; 85: 213–300. [DOI] [PubMed] [Google Scholar]
- 47. Meng X, Zhu Y, Di H, et al. Dose-response association of early-life antibiotic exposure and subsequent overweight or obesity in children: a meta-analysis of prospective studies. Obes Rev 2021; 22: e13321. [DOI] [PubMed] [Google Scholar]
- 48. Shao X, Ding X, Wang B, et al. Antibiotic exposure in early life increases risk of childhood obesity: a systematic review and meta-analysis. Front Endocrinol 2017; 8: 170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Rasmussen SH, Shrestha S, Bjerregaard LG, et al. Antibiotic exposure in early life and childhood overweight and obesity: a systematic review and meta-analysis. Diabetes Obes Metab 2018; 20: 1508–1514. [DOI] [PubMed] [Google Scholar]
- 50. Wan S, Guo M, Zhang T, et al. Impact of exposure to antibiotics during pregnancy and infancy on childhood obesity: a systematic review and meta-analysis. Obesity 2020; 28: 793–802. [DOI] [PubMed] [Google Scholar]
- 51. Backhed F, Roswall J, Peng Y, et al. Dynamics and stabilization of the human gut microbiome during the first year of life. Cell Host Microbe 2015; 17: 690–703. [DOI] [PubMed] [Google Scholar]
- 52. Turnbaugh PJ, Ley RE, Mahowald MA, et al. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature 2006; 444: 1027–1031. [DOI] [PubMed] [Google Scholar]
- 53. Principi N, Esposito S. Antibiotic administration and the development of obesity in children. Int J Antimicrob Agents 2016; 47: 171–177. [DOI] [PubMed] [Google Scholar]
- 54. Murugesan S, Nirmalkar K, Hoyo-Vadillo C, et al. Gut microbiome production of short-chain fatty acids and obesity in children. Eur J Clin Microbiol Infect Dis 2018; 37: 621–625. [DOI] [PubMed] [Google Scholar]
- 55. Joseph N, Vasodavan K, Saipudin NA, et al. Gut microbiota and short-chain fatty acids (SCFAs) profiles of normal and overweight school children in Selangor after probiotics administration. J Funct Foods 2019; 57: 103–111. [Google Scholar]
- 56. Abenavoli L, Scarpellini E, Colica C, et al. Gut Microbiota and obesity: a role for probiotics. Nutrients 2019; 11: 2690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Azad MB, Bridgman SL, Becker AB, et al. Infant antibiotic exposure and the development of childhood overweight and central adiposity. Int J Obes 2014; 38: 1290–1298. [DOI] [PubMed] [Google Scholar]
- 58. Schwartz BS, Pollak J, Bailey-Davis L, et al. Antibiotic use and childhood body mass index trajectory. Int J Obes 2016; 40: 615–621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Saari A, Virta LJ, Sankilampi U, et al. Antibiotic exposure in infancy and risk of being overweight in the first 24 months of life. Pediatrics 2015; 135: 617–626. [DOI] [PubMed] [Google Scholar]
- 60. Mir E, Panjabi C, Shah A. Impact of allergic rhinitis in school going children. Asia Pac Allergy 2012; 2: 93–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Chiu CY, Chan YL, Tsai MH, et al. Gut microbial dysbiosis is associated with allergen-specific IgE responses in young children with airway allergies. World Allergy Organ J 2019; 12: 100021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Simonyte Sjodin K, Hammarstrom ML, Rydén P, et al. Temporal and long-term gut microbiota variation in allergic disease: a prospective study from infancy to school age. Allergy 2019; 74: 176–185. [DOI] [PubMed] [Google Scholar]
- 63. Khan DA. Allergic rhinitis and asthma: epidemiology and common pathophysiology. Allergy Asthma Proc 2014; 35: 357–361. [DOI] [PubMed] [Google Scholar]
- 64. Nogacka AM, Salazar N, Arboleya S, et al. Early microbiota, antibiotics and health. Cell Mol Life Sci 2018; 75: 83–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Lin YC, Chen YC, Kuo CH, et al. Antibiotic exposure and asthma development in children with allergic rhinitis. J Microbiol Immunol Infect 2020; 53: 803–811. [DOI] [PubMed] [Google Scholar]
- 66. Gruber C, Keil T, Kulig M, et al. History of respiratory infections in the first 12 yr among children from a birth cohort. Pediatr Allergy Immunol 2008; 19: 505–512. [DOI] [PubMed] [Google Scholar]
- 67. Alkhater SA. Approach to the child with recurrent infections. J Family Community Med 2009; 16: 77–82. [PMC free article] [PubMed] [Google Scholar]
- 68. Langdon A, Crook N, Dantas G. The effects of antibiotics on the microbiome throughout development and alternative approaches for therapeutic modulation. Genome Med 2016; 8: 39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Kummeling I, Stelma FF, Dagnelie PC, et al. Early life exposure to antibiotics and the subsequent development of eczema, wheeze, and allergic sensitization in the first 2 years of life: the KOALA Birth Cohort Study. Pediatrics 2007; 119: e225–31. [DOI] [PubMed] [Google Scholar]
- 70. Hill CJ, Lynch DB, Murphy K, et al. Evolution of gut microbiota composition from birth to 24 weeks in the INFANTMET cohort. Microbiome 2017; 5: 4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Li L, Wang F, Liu Y, et al. Intestinal microbiota dysbiosis in children with recurrent respiratory tract infections. Microb Pathog 2019; 136: 103709. [DOI] [PubMed] [Google Scholar]
- 72. Bosch A, de Steenhuijsen Piters WAA, van Houten MA, et al. Maturation of the infant respiratory microbiota, environmental drivers, and health consequences: a prospective cohort study. Am J Respir Crit Care Med 2017; 196: 1582–1590. [DOI] [PubMed] [Google Scholar]
- 73. Robinson PFM, Pattaroni C, Cook J, et al. Lower airway microbiota associates with inflammatory phenotype in severe preschool wheeze. J Allergy Clin Immunol 2019; 143: 1607–1610. [DOI] [PubMed] [Google Scholar]
- 74. Bartlett JG. Clinical practice: antibiotic-associated diarrhea. N Engl J Med 2002; 346: 334–339. [DOI] [PubMed] [Google Scholar]
- 75. Wronowski MF, Kotowska M, Banasiuk M, et al. Bovine lactoferrin in the prevention of antibiotic-associated diarrhea in children: a randomized clinical trial. Front Pediatr 2021; 9: 675606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Zhang L, Zeng X, Guo D, et al. Early use of probiotics might prevent antibiotic-associated diarrhea in elderly (>65 years): a systematic review and meta-analysis. BMC Geriatr 2022; 22: 562. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Elseviers MM, Van Camp Y, Nayaert S, et al. Prevalence and management of antibiotic associated diarrhea in general hospitals. BMC Infect Dis 2015; 15: 129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Willing BP, Russell SL, Finlay BB. Shifting the balance: antibiotic effects on host-microbiota mutualism. Nat Rev Microbiol 2011; 9: 233–243. [DOI] [PubMed] [Google Scholar]
- 79. Yachha SK, Sarma MS, Mohan N, et al. Indian Academy of Pediatrics Consensus Guidelines for probiotic use in childhood diarrhea. Indian Pediatr 2022; 59: 543–551. [PubMed] [Google Scholar]
- 80. Hojsak I. Probiotics in children: what is the evidence? Pediatr Gastroenterol Hepatol Nutr 2017; 20: 139–146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Rupnik M, Wilcox MH, Gerding DN. Clostridium difficile infection: new developments in epidemiology and pathogenesis. Nat Rev Microbiol 2009; 7: 526–536. [DOI] [PubMed] [Google Scholar]
- 82. Bruno G, Rocco G, Zaccari P, et al. Helicobacter pylori infection and gastric dysbiosis: can probiotics administration be useful to treat this condition. Can J Infect Dis Med Microbiol 2018; 2018: 6237239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Hunt RH, Camilleri M, Crowe SE, et al. The stomach in health and disease. Gut 2015; 64: 1650–1668. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Klymiuk I, Bilgilier C, Stadlmann A, et al. The human gastric microbiome is predicated upon infection with Helicobacter pylori. Front Microbiol 2017; 8: 2508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Jernberg C, Lofmark S, Edlund C, et al. Long-term ecological impacts of antibiotic administration on the human intestinal microbiota. ISME J 2007; 1: 56–66. [DOI] [PubMed] [Google Scholar]
- 86. Lofmark S, Jernberg C, Jansson JK, et al. Clindamycin-induced enrichment and long-term persistence of resistant Bacteroides spp. J Antimicrob Chemother 2006; 58: 1160–1167. [DOI] [PubMed] [Google Scholar]
- 87. Dadgostar P. Antimicrobial resistance: implications and costs. Infect Drug Resist 2019; 12: 3903–3910. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Liu R, Hong J, Xu X, et al. Gut microbiome and serum metabolome alterations in obesity and after weight-loss intervention. Nat Med 2017; 23: 859–868. [DOI] [PubMed] [Google Scholar]
- 89. Ebm C, Cecconi M, Moran C, et al. Cost implication of antibiotic-associated diarrhoea and the financial impact of probiotic use in its prevention. Crit Care 2013; 17: P83. [Google Scholar]
- 90. Zhang D, Prabhu VS, Marcella SW. Attributable healthcare resource utilization and costs for patients with primary and recurrent Clostridium difficile infection in the United States. Clin Infect Dis 2018; 66: 1326–1332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Lahiri K, Jadhav K, Gahlowt P, et al. Bacillus clausii as an adjuvant therapy in acute childhood diarrhoea. IOSR JDMS 2015; 14: 74–76. [Google Scholar]
- 92. Laguipo AB. Dysbiosis diagnosis. News-Medical, 8 October 2018, https://www.news-medical.net/health/Dysbiosis-Diagnosis.aspx (accessed 21 March 21022)).
- 93. Budding AE, Grasman ME. IS-pro: high-throughput molecular fingerprinting of the intestinal microbiota. FASEB J 2010; 24: 4556–4564. [DOI] [PubMed] [Google Scholar]
- 94. Casen C, Vebo HC, Sekelja M, et al. Deviations in human gut microbiota: a novel diagnostic test for determining dysbiosis in patients with IBS or IBD. Aliment Pharmacol Ther 2015; 42: 71–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Gasbarrini A, Corazza GR, Gasbarrini G, et al. Methodology and indications of H2-breath testing in gastrointestinal diseases: the Rome Consensus Conference. Aliment Pharmacol Ther 2009; 29: 1–49. [DOI] [PubMed] [Google Scholar]
- 96. Reimer RA. Establishing the role of diet in the microbiota-disease axis. Nat Rev Gastroenterol Hepatol 2019; 16: 86–87. [DOI] [PubMed] [Google Scholar]
- 97. Lv W, Liu C, Ye C, et al. Structural modulation of gut microbiota during alleviation of antibiotic-associated diarrhea with herbal formula. Int J Biol Macromol 2017; 105: 1622–1629. [DOI] [PubMed] [Google Scholar]
- 98. Shi Y, Zhai Q, Li D, et al. Restoration of cefixime-induced gut microbiota changes by Lactobacillus cocktails and fructooligosaccharides in a mouse model. Microbiol Res 2017; 200: 14–24. [DOI] [PubMed] [Google Scholar]
- 99. Pascal M, Perez-Gordo M, Caballero T, et al. Microbiome and allergic diseases. Front Immunol 2018; 9: 1584. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Soldi S, Vasileiadis S, Lohner S, et al. Prebiotic supplementation over a cold season and during antibiotic treatment specifically modulates the gut microbiota composition of 3-6 year-old children. Benef Microbes 2019; 10: 253–263. [DOI] [PubMed] [Google Scholar]
- 101. Nicolucci AC, Hume MP, Martinez I, et al. Prebiotics reduce body fat and alter intestinal microbiota in children who are overweight or with obesity. Gastroenterology 2017; 153: 711–722. [DOI] [PubMed] [Google Scholar]
- 102. Hume MP, Nicolucci AC, Reimer RA. Prebiotic supplementation improves appetite control in children with overweight and obesity: a randomized controlled trial. Am J Clin Nutr 2017; 105: 790–799. [DOI] [PubMed] [Google Scholar]
- 103. Brunser O, Gotteland M, Cruchet S, et al. Effect of a milk formula with prebiotics on the intestinal microbiota of infants after an antibiotic treatment. Pediatr Res 2006; 59: 451–456. [DOI] [PubMed] [Google Scholar]
- 104. Khalighi A, Behdani R, Kouhestani S. Probiotics: a comprehensive review of their classification, mode of action and role in human nutrition. In: Rao V, Rao LG. (eds) Probiotics and prebiotics in human nutrition and health. London: InTechOpen, 2016, pp. 19–39. [Google Scholar]
- 105. Abbrescia A, Palese LL, Papa S, et al. Antibiotic sensitivity of Bacillus clausii strains in commercial preparation. Curr Med Chem 2014; 1: 102–110. [Google Scholar]
- 106. Ghelardi E, Celandroni F, Salvetti S, et al. Survival and persistence of Bacillus clausii in the human gastrointestinal tract following oral administration as spore-based probiotic formulation. J Appl Microbiol 2015; 119: 552–559. [DOI] [PubMed] [Google Scholar]
- 107. Jayanthi N, Ratna Sudha M. Bacillus clausii: the probiotic of choice in the treatment of diarrhoea. J Yoga Phys Ther 2015; 5: 4. [Google Scholar]
- 108. Yeşilyurt N, Yılmaz B, Ağagündüz D, et al. Involvement of probiotics and postbiotics in the immune system modulation. Biologics 2021; 1: 89–110. [Google Scholar]
- 109. Hempel S, Newberry SJ, Maher AR, et al. Probiotics for the prevention and treatment of antibiotic-associated diarrhea: a systematic review and meta-analysis. JAMA 2012; 307: 1959–1969. [DOI] [PubMed] [Google Scholar]
- 110. Johnston BC, Ma SS, Goldenberg JZ, et al. Probiotics for the prevention of Clostridium difficile-associated diarrhea: a systematic review and meta-analysis. Ann Intern Med 2012; 157: 878–888. [DOI] [PubMed] [Google Scholar]
- 111. Williams NT. Probiotics. Am J Health Syst Pharm 2010; 67: 449–458. [DOI] [PubMed] [Google Scholar]
- 112. Yesilyurt N, Yılmaz B, Agagunduz D, et al. Microbiome-based personalized nutrition as a result of the 4.0 technological revolution: a mini literature review. Process Biochem 2022; 121: 257–262. [Google Scholar]
- 113. Nista EC, Candelli M, Cremonini F, et al. Bacillus clausii therapy to reduce side-effects of anti-Helicobacter pylori treatment: randomized, double-blind, placebo controlled trial. Aliment Pharmacol Ther 2004; 20: 1181–1188. [DOI] [PubMed] [Google Scholar]
- 114. Korpela K, Salonen A, Virta LJ, et al. Lactobacillus rhamnosus GG intake modifies preschool children’s intestinal microbiota, alleviates penicillin-associated changes, and reduces antibiotic use. PLoS ONE 2016; 11: e0154012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Brusaferro A, Cozzali R, Orabona C, et al. Is it time to use probiotics to prevent or treat obesity? Nutrients 2018; 10: 1613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Wang Y, Li X, Ge T, et al. Probiotics for prevention and treatment of respiratory tract infections in children: a systematic review and meta-analysis of randomized controlled trials. Medicine 2016; 95: e4509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Hojsak I, Snovak N, Abdovic S, et al. Lactobacillus GG in the prevention of gastrointestinal and respiratory tract infections in children who attend day care centers: a randomized, double-blind, placebo-controlled trial. Clin Nutr 2010; 29: 312–316. [DOI] [PubMed] [Google Scholar]
- 118. Destura RV, Genuino ML, De Castro JA. Efficacy and safety of Bacillus clausii in preventing antibiotic-associated diarrhea among Filipino infants and children: a multi-center, randomized, open-label clinical trial (NCT 00447161), 2008. [Google Scholar]
- 119. Choueiry E, Kamel R, Chacar H, et al. Lebanese registry in the management of antibiotic associated diarrhea in children: observational study in daily practice. Int J Clin Pediatr 2017; 6: 8–19. [Google Scholar]
- 120. McFarland LV, Goh S. Preventing pediatric antibiotic-associated diarrhea and Clostridium difficile infections with probiotics: a meta-analysis. World J Meta-Anal 2013; 1: 102–120. [Google Scholar]
- 121. Gao XW, Mubasher M, Fang CY, et al. Dose-response efficacy of a proprietary probiotic formula of Lactobacillus acidophilus CL1285 and Lactobacillus casei LBC80R for antibiotic-associated diarrhea and Clostridium difficile-associated diarrhea prophylaxis in adult patients. Am J Gastroenterol 2010; 105: 1636–1641. [DOI] [PubMed] [Google Scholar]
- 122. Chatterjee S, Kar P, Das T, et al. Randomised placebo-controlled double blind multicentric trial on efficacy and safety of Lactobacillus acidophilus LA-5 and Bifidobacterium BB-12 for prevention of antibiotic-associated diarrhoea. J Assoc Physicians India 2013; 61: 708–712. [PubMed] [Google Scholar]
- 123. Evans M, Salewski RP, Christman MC, et al. Effectiveness of Lactobacillus helveticus and Lactobacillus rhamnosus for the management of antibiotic-associated diarrhoea in healthy adults: a randomised, double-blind, placebo-controlled trial. Br J Nutr 2016; 116: 94–103. [DOI] [PubMed] [Google Scholar]
- 124. Sampalis J, Psaradellis E, Rampakakis E. Efficacy of BIO K+ CL1285 in the reduction of antibiotic-associated diarrhea: a placebo controlled double-blind randomized, multi-center study. Arch Med Sci 2010; 6: 56–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Syngai GG, Gopi R, Bharali R, et al. Probiotics: the versatile functional food ingredients. J Food Sci Technol 2016; 53: 921–933. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Guo Q, Goldenberg JZ, Humphrey C, et al. Probiotics for the prevention of pediatric antibiotic-associated diarrhea. Cochrane Database Syst Rev 2019; 4: CD004827. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Szajewska H, Kolodziej M. Systematic review with meta-analysis: Lactobacillus rhamnosus GG in the prevention of antibiotic-associated diarrhoea in children and adults. Aliment Pharmacol Ther 2015; 42: 1149–1157. [DOI] [PubMed] [Google Scholar]
- 128. Szajewska H, Kolodziej M. Systematic review with meta-analysis: Saccharomyces boulardii in the prevention of antibiotic-associated diarrhoea. Aliment Pharmacol Ther 2015; b42: 793–801. [DOI] [PubMed] [Google Scholar]
- 129. De Castro JA, Kesavelu D, Lahiri KR, et al. Recommendations for the adjuvant use of the poly-antibiotic-resistant probiotic Bacillus clausii (O/C, SIN, N/R, T) in acute, chronic, and antibiotic-associated diarrhea in children: consensus from Asian experts. Trop Dis Travel Med Vaccines 2020; 6: 21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Goldenberg JZ, Yap C, Lytvyn L, et al. Probiotics for the prevention of Clostridium difficile-associated diarrhea in adults and children. Cochrane Database Syst Rev 2017; 12: CD006095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Szajewska H, Canani RB, Guarino A, et al. Probiotics for the prevention of antibiotic-associated diarrhea in children. J Pediatr Gastroenterol Nutr 2016; 62: 495–506. [DOI] [PubMed] [Google Scholar]
- 132. Alvi S, Javeed A, Akhtar B, et al. Probiotics for cure of Helicobacter pylori infection: a review. Int J Food Prop 2017; 20: 2215–2222. [Google Scholar]
- 133. Plomer M, Iii, Perez M, Greifenberg DM. Effect of Bacillus clausii capsules in reducing adverse effects associated with Helicobacter pylori eradication therapy: a randomized, double-blind, controlled trial. Infect Dis Ther 2020; 9: 867–878. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Homan M, Orel R. Are probiotics useful in Helicobacter pylori eradication? World J Gastroenterol 2015; 21: 10644–10653. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Dang Y, Reinhardt JD, Zhou X, et al. The effect of probiotics supplementation on Helicobacter pylori eradication rates and side effects during eradication therapy: a meta-analysis. PLoS ONE 2014; 9: e111030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Katelaris P, Hunt R, Bazzoli F, et al. World Gastroenterology Organization global guidelines: Helicobacter pylori 2021, https://www.worldgastroenterology.org/guidelines/helicobacter-pylori (accessed 15 June 2022). [DOI] [PubMed]
- 137. Gour D, Toppo M, Pal DK, et al. Prescription audit of treatment of diarrhoea and pneumonia amongst paediatrician in Bhopal. Indian J Community Med 2020; 45: 554–557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Baidya S, Hazra A, Datta S, et al. A study of antimicrobial use in children admitted to pediatric medicine ward of a tertiary care hospital. Indian J Pharmacol 2017; 49: 10–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Sudha MR, Jayanthi N, Pandey DC, et al. Bacillus clausii UBBC-07 reduces severity of diarrhoea in children under 5 years of age: a double blind placebo controlled study. Beneficial Microbes 2019; 10: 149–154. [DOI] [PubMed] [Google Scholar]
- 140. Bhat S, Shreekrishna GN, Savio CD. Efficacy of probiotics in acute diarrhoea in children. Int J Contemp Pediatr 2018; 5: 1646–1650. [Google Scholar]
- 141. Vidjeadevan D, Vinoth S, Ramesh S. Role of Saccharomyces boulardii and Bacillus clausii in reducing the duration of diarrhea: a three-armed randomised controlled trial. Int J Contemp Pediatr 2018; 5: 1811–1814. [Google Scholar]
- 142. Das S, Gupta PK, Das RR. Efficacy and safety of Saccharomyces boulardii in acute rotavirus diarrhea: double blind randomized controlled trial from a developing country. J Trop Pediatr 2016: 1–7. [DOI] [PubMed] [Google Scholar]
- 143. Lahiri K, D’Souza J, Gahlowt P. Beneficial role of probiotic in acute childhood diarrhoea. J Harmoniz Res Med and Health Sci 2015; a2: 26–30. [Google Scholar]
- 144. Shireen A, Aneesh M. Knowledge and consumption of probiotics and prebiotics in India: a narrative review. Int J Community Med Public Health 2021; 8: 5119–5126. [Google Scholar]
- 145. Lakshmy G, Seetha Devi B, Ramesh B. The blooming prospects of probiotic products in India. Int J Recent Technol Eng 2018; 4: 253–258. [Google Scholar]
- 146. Raghuwanshi S, Misra S, Sharma R, et al. Indian perspective for probiotics: a review. Indian J Dairy Sci 2015; 68: 195–205. [Google Scholar]
- 147. Kesavelu D, Rohit A, Karunasagar I. Composition and laboratory correlation of commercial probiotics in India. Cureus 2020; 12: e11334. [DOI] [PMC free article] [PubMed] [Google Scholar]